1
|
Bayram Z, Akcabag E, Ozbey G, Nacitarhan C, Ozdem S, Turkay C, Ozdem SS. THE effect of P2X7 receptor activation on functional responses of human left internal mammary artery. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03411-1. [PMID: 39225832 DOI: 10.1007/s00210-024-03411-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
The Purinoreceptor 7 (P2X7R) has become a promising drug target in many cardiovascular diseases, including coronary artery disease, since prolonged activation of P2X7R could promote vascular dysfunction, atherosclerosis, and thrombosis. Thus, we aimed to study the effects of P2X7R activation on vascular relaxation responses of the human left internal mammary artery (LIMA). Sections of redundant human LIMA were cut into 3-mm wide rings,, suspended in 20-mL organ baths containing physiologic salt solution, and attached to an isometric force transducer connected to a computer-based data acquisition system. Long-term (60 min) incubation with specific P2X7R agonist Bz-ATP caused significant reductions in relaxation responses of LIMA to ATP and acetylcholine, which were reversed by selective P2X7R antagonists Brilliant Blue G or AZ11645373, whereas there were no changes in relaxation responses to endothelium-independent vasodilators isoprenaline, cAMP analog 8-Br-cAMP, and nitric oxide donor sodium nitroprusside. The impairment in relaxant responses of LIMA to endothelium-dependent vasodilators following activation of P2X7R for the long-term may contribute to postoperative LIMA vasospasm and hypertension. Modulation of P2X7R activity with selective agents may represent a new potential therapeutic approach in patients undergoing coronary artery bypass grafting surgery.
Collapse
Affiliation(s)
- Zeliha Bayram
- Turkish Medicines and Medical Devices Agency, Ankara, Turkey
| | - Esra Akcabag
- Department of Medical Pharmacology, Akdeniz University Medical Faculty, Dumlupinar Avenue, 07070, Antalya, Turkey
| | - Gul Ozbey
- Department of Medical Pharmacology, Akdeniz University Medical Faculty, Dumlupinar Avenue, 07070, Antalya, Turkey
| | - Cahit Nacitarhan
- Department of Medical Pharmacology, Akdeniz University Medical Faculty, Dumlupinar Avenue, 07070, Antalya, Turkey
| | - Sebahat Ozdem
- Department of Medical Biochemistry, Akdeniz University Medical Faculty, Antalya, Turkey
| | - Cengiz Turkay
- Department of Cardiovascular Surgery, Akdeniz University Medical Faculty, Antalya, Turkey
| | - Sadi S Ozdem
- Department of Medical Pharmacology, Akdeniz University Medical Faculty, Dumlupinar Avenue, 07070, Antalya, Turkey.
| |
Collapse
|
2
|
Ye T, Yang J, Liu Z, Yu Y, Zhang C, Guo Y, Yu F, Zhou Y, Song Z, Shi J, Wang L, Yang B, Wang X. Inhibition of the P2X7 receptor prevents atrial proarrhythmic remodeling in experimental post-operative atrial fibrillation. Int Immunopharmacol 2024; 129:111536. [PMID: 38320354 DOI: 10.1016/j.intimp.2024.111536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 02/08/2024]
Abstract
BACKGROUND Post-operative atrial fibrillation (POAF) is a common complication in patients undergoing cardiac surgery. The purinergic receptor P2X7 (P2X7R) is involved in some cardiovascular diseases, whereas its effects on atrial fibrillation (AF) are unclear. OBJECTIVE This study was to assess the effect of P2X7R on atrial arrhythmogenic remodeling in the rat model of sterile pericarditis (SP). METHODS Male Sprague-Dawley (SD) rats were used to induce the SP model. Electrocardiogram, atrial electrophysiological protocol, histology, mRNA sequencing, real-time quantitative PCR, western blot, and Elisa assay were performed. RESULTS SP significantly up-regulated P2X7R expression; increased AF susceptibility; reduced the protein expression of ion channels including Nav1.5, Cav1.2, Kv4.2, Kv4.3, and Kv1.5; caused atrial fibrosis; increased norepinephrine (NE) level in plasma; promoted the production of inflammatory cytokines such as TNF-α, IL-1β, and IL-6; increased the accumulation of immune cells (CD68- and MPO- positive cells); and activated NLRP3 inflammasome signaling pathway. P2X7R antagonist Brilliant Blue G (BBG) mitigated SP-induced alterations. The mRNA sequencing demonstrated that BBG prevented POAF mainly by regulating the immune system. In addition, another selective P2X7R antagonist A740003, and IL-1R antagonist anakinra also reduced AF inducibility in the SP model. CONCLUSIONS P2X7R inhibition prevents SP-induced atrial proarrhythmic remodeling, which is closely associated with the improvement of inflammatory changes, ion channel expression, atrial fibrosis, and sympathetic activation. The findings point to P2X7R inhibition as a promising target for AF (particularly POAF) and perhaps other conditions.
Collapse
Affiliation(s)
- Tianxin Ye
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Jinxiu Yang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Zhangchi Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Yi Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Cui Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Yan Guo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Fangcong Yu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yunping Zhou
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Zhuonan Song
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Jiaran Shi
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Longbo Wang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China.
| | - Xingxiang Wang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
3
|
Zabłocki K, Górecki DC. The Role of P2X7 Purinoceptors in the Pathogenesis and Treatment of Muscular Dystrophies. Int J Mol Sci 2023; 24:ijms24119434. [PMID: 37298386 DOI: 10.3390/ijms24119434] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Muscular dystrophies are inherited neuromuscular diseases, resulting in progressive disability and often affecting life expectancy. The most severe, common types are Duchenne muscular dystrophy (DMD) and Limb-girdle sarcoglycanopathy, which cause advancing muscle weakness and wasting. These diseases share a common pathomechanism where, due to the loss of the anchoring dystrophin (DMD, dystrophinopathy) or due to mutations in sarcoglycan-encoding genes (LGMDR3 to LGMDR6), the α-sarcoglycan ecto-ATPase activity is lost. This disturbs important purinergic signaling: An acute muscle injury causes the release of large quantities of ATP, which acts as a damage-associated molecular pattern (DAMP). DAMPs trigger inflammation that clears dead tissues and initiates regeneration that eventually restores normal muscle function. However, in DMD and LGMD, the loss of ecto-ATPase activity, that normally curtails this extracellular ATP (eATP)-evoked stimulation, causes exceedingly high eATP levels. Thus, in dystrophic muscles, the acute inflammation becomes chronic and damaging. The very high eATP over-activates P2X7 purinoceptors, not only maintaining the inflammation but also tuning the potentially compensatory P2X7 up-regulation in dystrophic muscle cells into a cell-damaging mechanism exacerbating the pathology. Thus, the P2X7 receptor in dystrophic muscles is a specific therapeutic target. Accordingly, the P2X7 blockade alleviated dystrophic damage in mouse models of dystrophinopathy and sarcoglycanopathy. Therefore, the existing P2X7 blockers should be considered for the treatment of these highly debilitating diseases. This review aims to present the current understanding of the eATP-P2X7 purinoceptor axis in the pathogenesis and treatment of muscular dystrophies.
Collapse
Affiliation(s)
- Krzysztof Zabłocki
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Dariusz C Górecki
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK
| |
Collapse
|
4
|
Zhuang Y, Yu ML, Lu SF. Purinergic signaling in myocardial ischemia-reperfusion injury. Purinergic Signal 2023; 19:229-243. [PMID: 35254594 PMCID: PMC9984618 DOI: 10.1007/s11302-022-09856-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 02/18/2022] [Indexed: 10/18/2022] Open
Abstract
Purines and their derivatives, extensively distributed in the body, act as a class of extracellular signaling molecules via a rich array of receptors, also known as purinoceptors (P1, P2X, and P2Y). They mediate multiple intracellular signal transduction pathways and participate in various physiological and pathological cell behaviors. Since the function in myocardial ischemia-reperfusion injury (MIRI), this review summarized the involvement of purinergic signal transduction in diversified pathological processes, including energy metabolism disorder, oxidative stress injury, calcium overload, inflammatory immune response, platelet aggregation, coronary vascular dysfunction, and cell necrosis and apoptosis. Moreover, increasing evidence suggests that purinergic signaling also mediates the prevention and treatment of MIRI, such as ischemic conditioning, pharmacological intervention, and some other therapies. In conclusion, this review exhibited that purinergic signaling mediates the complex processes of MIRI which shows its promising application and prospecting in the future.
Collapse
Affiliation(s)
- Yi Zhuang
- College of Acupuncture and Tuina, Nanjing University of Chinese Medicine, 138 Xian-lin Avenue, Qixia District, Nanjing, 210023, Jiangsu Province, China
| | - Mei-Ling Yu
- College of Acupuncture and Tuina, Nanjing University of Chinese Medicine, 138 Xian-lin Avenue, Qixia District, Nanjing, 210023, Jiangsu Province, China
| | - Sheng-Feng Lu
- College of Acupuncture and Tuina, Nanjing University of Chinese Medicine, 138 Xian-lin Avenue, Qixia District, Nanjing, 210023, Jiangsu Province, China. .,Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
5
|
Onódi Z, Koch S, Rubinstein J, Ferdinandy P, Varga ZV. Drug repurposing for cardiovascular diseases: New targets and indications for probenecid. Br J Pharmacol 2023; 180:685-700. [PMID: 36484549 DOI: 10.1111/bph.16001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 11/12/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
The available pharmacological options in the management of cardiovascular diseases such as ischaemic heart disease and subsequent heart failure are effective in slowing the progression of this condition. However, the long-term prognosis is still poor, raising the demand for new therapeutic strategies. Drug repurposing is a time- and cost-effective drug development strategy that offers approved and abandoned drugs a new chance for new indications. Recently, drugs used for the management of gout-related inflammation such as canakinumab or colchicine have been considered for drug repurposing in cardiovascular indications. The old uricosuric drug, probenecid, has been identified as a novel therapeutic option in the management of specific cardiac diseases as well. Probenecid can modulate myocardial contractility and vascular tone and exerts anti-inflammatory properties. The mechanisms behind these beneficial effects might be related inhibition of inflammasomes, and to modulation purinergic-pannexin-1 signalling and TRPV2 channels, which are recently identified molecular targets of probenecid. In this review, we provide an overview on repurposing probenecid for ischaemic heart disease and subsequent heart failure by summarizing the related experimental and clinical data and propose its potential repurposing to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Zsófia Onódi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,HCEMM-SU Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary.,MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, Budapest, Hungary
| | - Sheryl Koch
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Jack Rubinstein
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,HCEMM-SU Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary.,MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, Budapest, Hungary
| |
Collapse
|
6
|
Sudi S, Thomas FM, Daud SK, Ag Daud DM, Sunggip C. The Pleiotropic Role of Extracellular ATP in Myocardial Remodelling. Molecules 2023; 28:molecules28052102. [PMID: 36903347 PMCID: PMC10004151 DOI: 10.3390/molecules28052102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 03/12/2023] Open
Abstract
Myocardial remodelling is a molecular, cellular, and interstitial adaptation of the heart in response to altered environmental demands. The heart undergoes reversible physiological remodelling in response to changes in mechanical loading or irreversible pathological remodelling induced by neurohumoral factors and chronic stress, leading to heart failure. Adenosine triphosphate (ATP) is one of the potent mediators in cardiovascular signalling that act on the ligand-gated (P2X) and G-protein-coupled (P2Y) purinoceptors via the autocrine or paracrine manners. These activations mediate numerous intracellular communications by modulating the production of other messengers, including calcium, growth factors, cytokines, and nitric oxide. ATP is known to play a pleiotropic role in cardiovascular pathophysiology, making it a reliable biomarker for cardiac protection. This review outlines the sources of ATP released under physiological and pathological stress and its cell-specific mechanism of action. We further highlight a series of cardiovascular cell-to-cell communications of extracellular ATP signalling cascades in cardiac remodelling, which can be seen in hypertension, ischemia/reperfusion injury, fibrosis, hypertrophy, and atrophy. Finally, we summarize current pharmacological intervention using the ATP network as a target for cardiac protection. A better understanding of ATP communication in myocardial remodelling could be worthwhile for future drug development and repurposing and the management of cardiovascular diseases.
Collapse
Affiliation(s)
- Suhaini Sudi
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
| | - Fiona Macniesia Thomas
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
| | - Siti Kadzirah Daud
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
| | - Dayang Maryama Ag Daud
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
- Health through Exercise and Active Living (HEAL) Research Unit, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
| | - Caroline Sunggip
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
- Borneo Medical and Health Research Centre, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
- Correspondence:
| |
Collapse
|
7
|
Jiang Y, Lin J, Zheng H, Zhu P. The Role of Purinergic Signaling in Heart Transplantation. Front Immunol 2022; 13:826943. [PMID: 35529844 PMCID: PMC9069525 DOI: 10.3389/fimmu.2022.826943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/22/2022] [Indexed: 11/13/2022] Open
Abstract
Heart transplantation remains the optimal treatment option for patients with end-stage heart disease. Growing evidence demonstrates that purinergic signals mediated by purine nucleotides and nucleosides play vital roles in heart transplantation, especially in the era of ischemia-reperfusion injury (IRI) and allograft rejection. Purinergic signaling consists of extracellular nucleotides and nucleosides, ecto-enzymes, and cell surface receptors; it participates in the regulation of many physiological and pathological processes. During transplantation, excess adenosine triphosphate (ATP) levels are released from damaged cells, and driver detrimental inflammatory responses largely via purinergic P2 receptors. Ecto-nucleosidases sequentially dephosphorylate extracellular ATP to ADP, AMP, and finally adenosine. Adenosine exerts a cardioprotective effect by its anti-inflammatory, antiplatelet, and vasodilation properties. This review focused on the role of purinergic signaling in IRI and rejection after heart transplantation, as well as the clinical applications and prospects of purinergic signaling.
Collapse
Affiliation(s)
| | | | | | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
8
|
Signaling pathways and targeted therapy for myocardial infarction. Signal Transduct Target Ther 2022; 7:78. [PMID: 35273164 PMCID: PMC8913803 DOI: 10.1038/s41392-022-00925-z] [Citation(s) in RCA: 258] [Impact Index Per Article: 129.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/28/2022] [Accepted: 02/08/2022] [Indexed: 02/07/2023] Open
Abstract
Although the treatment of myocardial infarction (MI) has improved considerably, it is still a worldwide disease with high morbidity and high mortality. Whilst there is still a long way to go for discovering ideal treatments, therapeutic strategies committed to cardioprotection and cardiac repair following cardiac ischemia are emerging. Evidence of pathological characteristics in MI illustrates cell signaling pathways that participate in the survival, proliferation, apoptosis, autophagy of cardiomyocytes, endothelial cells, fibroblasts, monocytes, and stem cells. These signaling pathways include the key players in inflammation response, e.g., NLRP3/caspase-1 and TLR4/MyD88/NF-κB; the crucial mediators in oxidative stress and apoptosis, for instance, Notch, Hippo/YAP, RhoA/ROCK, Nrf2/HO-1, and Sonic hedgehog; the controller of myocardial fibrosis such as TGF-β/SMADs and Wnt/β-catenin; and the main regulator of angiogenesis, PI3K/Akt, MAPK, JAK/STAT, Sonic hedgehog, etc. Since signaling pathways play an important role in administering the process of MI, aiming at targeting these aberrant signaling pathways and improving the pathological manifestations in MI is indispensable and promising. Hence, drug therapy, gene therapy, protein therapy, cell therapy, and exosome therapy have been emerging and are known as novel therapies. In this review, we summarize the therapeutic strategies for MI by regulating these associated pathways, which contribute to inhibiting cardiomyocytes death, attenuating inflammation, enhancing angiogenesis, etc. so as to repair and re-functionalize damaged hearts.
Collapse
|
9
|
Sarti AC, Vultaggio-Poma V, Falzoni S, Missiroli S, Giuliani AL, Boldrini P, Bonora M, Faita F, Di Lascio N, Kusmic C, Solini A, Novello S, Morari M, Rossato M, Wieckowski MR, Giorgi C, Pinton P, Di Virgilio F. Mitochondrial P2X7 Receptor Localization Modulates Energy Metabolism Enhancing Physical Performance. FUNCTION (OXFORD, ENGLAND) 2021; 2:zqab005. [PMID: 35330818 PMCID: PMC8788778 DOI: 10.1093/function/zqab005] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 02/06/2023]
Abstract
Basal expression of the P2X7 receptor (P2X7R) improves mitochondrial metabolism, Adenosine 5'-triphosphate (ATP) synthesis, and overall fitness of immune and non-immune cells. We investigated P2X7R contribution to energy metabolism and subcellular localization in fibroblasts (mouse embryo fibroblasts and HEK293 human fibroblasts), mouse microglia (primary brain microglia, and the N13 microglia cell line), and heart tissue. The P2X7R localizes to mitochondria, and its lack (1) decreases basal respiratory rate, ATP-coupled respiration, maximal uncoupled respiration, resting mitochondrial potential, mitochondrial matrix Ca2+ level, (2) modifies expression pattern of oxidative phosphorylation enzymes, and (3) severely affects cardiac performance. Hearts from P2rx7-deleted versus wild-type mice are larger, heart mitochondria smaller, and stroke volume, ejection fraction, fractional shortening, and cardiac output, are significantly decreased. Accordingly, the physical fitness of P2X7R-null mice is severely reduced. Thus, the P2X7R is a key modulator of mitochondrial energy metabolism and a determinant of physical fitness.
Collapse
Affiliation(s)
- Alba Clara Sarti
- Department of Medical Sciences, University of
Ferrara, Ferrara 44121, Italy
| | | | - Simonetta Falzoni
- Department of Medical Sciences, University of
Ferrara, Ferrara 44121, Italy
| | - Sonia Missiroli
- Department of Medical Sciences, University of
Ferrara, Ferrara 44121, Italy
| | - Anna Lisa Giuliani
- Department of Medical Sciences, University of
Ferrara, Ferrara 44121, Italy
| | - Paola Boldrini
- Center of Electronic Microscopy, University of
Ferrara, Ferrara 44121, Italy
| | - Massimo Bonora
- Department of Medical Sciences, University of
Ferrara, Ferrara 44121, Italy
| | - Francesco Faita
- Institute of Clinical Physiology, National Research
Council, Pisa 56124, Italy
| | - Nicole Di Lascio
- Institute of Clinical Physiology, National Research
Council, Pisa 56124, Italy
| | - Claudia Kusmic
- Institute of Clinical Physiology, National Research
Council, Pisa 56124, Italy
| | - Anna Solini
- Department of Surgical, Medical, Molecular, and
Critical Area Pathology, University of Pisa, Pisa 56124, Italy
| | - Salvatore Novello
- Department of Biomedical and Specialty Surgical
Sciences, University of Ferrara, Ferrara 44121, Italy
| | - Michele Morari
- Department of Biomedical and Specialty Surgical
Sciences, University of Ferrara, Ferrara 44121, Italy
| | - Marco Rossato
- Department of Medicine, University of
Padova, Padova 35128, Italy
| | | | - Carlotta Giorgi
- Department of Medical Sciences, University of
Ferrara, Ferrara 44121, Italy
| | - Paolo Pinton
- Department of Medical Sciences, University of
Ferrara, Ferrara 44121, Italy
| | - Francesco Di Virgilio
- Department of Medical Sciences, University of
Ferrara, Ferrara 44121, Italy,Address correspondence to F.D.V. (e-mail:
)
| |
Collapse
|
10
|
Woo SH, Trinh TN. P2 Receptors in Cardiac Myocyte Pathophysiology and Mechanotransduction. Int J Mol Sci 2020; 22:ijms22010251. [PMID: 33383710 PMCID: PMC7794727 DOI: 10.3390/ijms22010251] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/22/2020] [Accepted: 12/22/2020] [Indexed: 12/30/2022] Open
Abstract
ATP is a major energy source in the mammalian cells, but it is an extracellular chemical messenger acting on P2 purinergic receptors. A line of evidence has shown that ATP is released from many different types of cells including neurons, endothelial cells, and muscle cells. In this review, we described the distribution of P2 receptor subtypes in the cardiac cells and their physiological and pathological roles in the heart. So far, the effects of external application of ATP or its analogues, and those of UTP on cardiac contractility and rhythm have been reported. In addition, specific genetic alterations and pharmacological agonists and antagonists have been adopted to discover specific roles of P2 receptor subtypes including P2X4-, P2X7-, P2Y2- and P2Y6-receptors in cardiac cells under physiological and pathological conditions. Accumulated data suggest that P2X4 receptors may play a beneficial role in cardiac muscle function, and that P2Y2- and P2Y6-receptors can induce cardiac fibrosis. Recent evidence further demonstrates P2Y1 receptor and P2X4 receptor as important mechanical signaling molecules to alter membrane potential and Ca2+ signaling in atrial myocytes and their uneven expression profile between right and left atrium.
Collapse
|
11
|
Shokoples BG, Paradis P, Schiffrin EL. P2X7 Receptors: An Untapped Target for the Management of Cardiovascular Disease. Arterioscler Thromb Vasc Biol 2020; 41:186-199. [PMID: 32998520 PMCID: PMC7752223 DOI: 10.1161/atvbaha.120.315116] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chronic low-grade inflammation contributes to the development of several diseases, including cardiovascular disease. Adequate strategies to target inflammation in cardiovascular disease are in their infancy and remain an avenue of great interest. The purinergic receptor P2X7 is a ubiquitously expressed receptor that predominately mediates inflammation and cellular death. P2X7 is a ligand-gated cation channel that is activated in response to high concentrations of extracellular ATP, triggering the assembly and activation of the NLRP3 (nuclear oligomerization domain like receptor family pyrin domain containing 3) inflammasome and subsequent release of proinflammatory cytokines IL (interleukin)-1β and IL-18. Increased P2X7 activation and IL-1β and IL-18 concentrations have been implicated in the development of many cardiovascular conditions including hypertension, atherosclerosis, ischemia/reperfusion injury, and heart failure. P2X7 receptor KO (knockout) mice exhibit a significant attenuation of the inflammatory response, which corresponds with reduced disease severity. P2X7 antagonism blunts blood pressure elevation in hypertension and progression of atherosclerosis in animal models. IL-1β and IL-18 inhibition has shown efficacy in clinical trials reducing major adverse cardiac events, including myocardial infarction, and heart failure. With several P2X7 antagonists available with proven safety margins, P2X7 antagonism could represent an untapped potential for therapeutic intervention in cardiovascular disorders.
Collapse
Affiliation(s)
- Brandon G. Shokoples
- Vascular and Hypertension Research Unit, Lady Davis Institute for Medical Research (B.G.S., P.P., E.L.S.), Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Pierre Paradis
- Vascular and Hypertension Research Unit, Lady Davis Institute for Medical Research (B.G.S., P.P., E.L.S.), Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Ernesto L. Schiffrin
- Vascular and Hypertension Research Unit, Lady Davis Institute for Medical Research (B.G.S., P.P., E.L.S.), Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, Quebec, Canada
- Department of Medicine (E.L.S.), Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
12
|
Arkhipov SN, Pavlov TS. ATP release into ADPKD cysts via pannexin-1/P2X7 channels decreases ENaC activity. Biochem Biophys Res Commun 2019; 513:166-171. [PMID: 30952430 PMCID: PMC6475605 DOI: 10.1016/j.bbrc.2019.03.177] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 03/26/2019] [Indexed: 12/30/2022]
Abstract
Genetic predisposition is necessary for polycystic kidney disease (PKD) initiation, although there are other, incompletely identified downstream processes that are required for cyst growth. Their characterization may provide a unique opportunity for clinical interventions. One of the poorly studied phenomena in PKD is high ATP content in cysts. Unfortunately, neither origins of uncontrolled ATP release, nor consequences of abnormal purinergic signaling in relation to epithelial transport are well explored in the polycystic kidney. We tested the distribution of pannexin-1 (Panx1) and P2X7, two proteins potentially involved in ATP release, in the kidneys of the Pkd1RC/RC mice, a model of autosomal dominant PKD (ADPKD). Abundances of both proteins were abnormally increased in the cyst lining cells compared to non-dilated collecting ducts. To establish if pannexin-1 contributes to ATP release in the collecting ducts (CD), we measured luminal accumulation of ATP in M1 cell renal CD monolayers, and found that treatment with probenecid, a Panx1 blocker, prevents ATP release. Single channel patch clamp analysis of polarized M1 cells revealed that apical stimulation of P2X receptors with αβ-MeATP acutely reduces ENaC activity. We conclude that in ADPKD progression, an abnormal hyperexpression of both PANX1 and P2RX7 occurs in the cyst lining epithelial cells. High abundance of both proteins is not typical for non-dilated CDs but, when it happens in cysts, pannexin1/P2X7 cooperation elevates ATP release into the luminal space. High ATP level is a pathogenic factor facilitating cystogenesis by reducing ENaC-mediated reabsorption from the lumen.
Collapse
Affiliation(s)
- Sergey N Arkhipov
- Division of Hypertension and Vascular Research, Henry Ford Health System, Detroit, MI, USA
| | - Tengis S Pavlov
- Division of Hypertension and Vascular Research, Henry Ford Health System, Detroit, MI, USA.
| |
Collapse
|
13
|
Guerra Martinez C. P2X7 receptor in cardiovascular disease: The heart side. Clin Exp Pharmacol Physiol 2019; 46:513-526. [PMID: 30834550 DOI: 10.1111/1440-1681.13079] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 02/21/2019] [Accepted: 02/28/2019] [Indexed: 01/10/2023]
Abstract
The P2X7 receptor is a ligand-gated purinergic receptor activated by extracellular ATP. The receptor is highly expressed in immune cells and in the brain, and, upon activation, the P2X7 receptor allows a cation flux, leading to the distinct activation of intracellular signalling pathways as the secretion of pro-inflammatory cytokines, and modulation of cell survival. Through these molecular mechanisms, P2X7 is known to play important roles in physiology and pathophysiology of a wide spectrum of diseases, including cancer, inflammatory diseases, neurological, respiratory and more recently cardiovascular diseases. Recent studies demonstrated that the P2X7 could modulate the assembly of the NLRP3 inflammasome, leading to the secretion of pro-inflammatory factors and worsen the cardiac disease phenotypes. This review discusses the critical molecular function of P2X7 in the modulation of the onset, progression and resolution of cardiovascular diseases and analyses the putative future use of P2X7-based therapies that modulate the IL-1β secretion arm and direct P2X7 antagonists.
Collapse
Affiliation(s)
- Camila Guerra Martinez
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas
| |
Collapse
|
14
|
Novielli-Kuntz NM, Jelen M, Barr K, DeLalio LJ, Feng Q, Isakson BE, Gros R, Laird DW. Ablation of both Cx40 and Panx1 results in similar cardiovascular phenotypes exhibited in Cx40 knockout mice. Biosci Rep 2019; 39:BSR20182350. [PMID: 30745457 PMCID: PMC6393227 DOI: 10.1042/bsr20182350] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/10/2019] [Accepted: 02/05/2019] [Indexed: 11/30/2022] Open
Abstract
Connexins (Cxs) and pannexins (Panxs) are highly regulated large-pore channel-forming proteins that participate in cellular communication via small molecular exchange with the extracellular microenvironment, or in the case of connexins, directly between cells. Given the putative functional overlap between single membrane-spanning connexin hemichannels and Panx channels, and cardiovascular system prevalence, we generated the first Cx40-/-Panx1-/- mouse with the anticipation that this genetic modification would lead to a severe cardiovascular phenotype. Mice null for both Cx40 and Panx1 produced litter sizes and adult growth progression similar to wild-type (WT), Cx40-/- and Panx1-/- mice. Akin to Cx40-/- mice, Cx40-/-Panx1-/- mice exhibited cardiac hypertrophy and elevated systolic, diastolic, and mean arterial blood pressure compared with WT and Panx1-/- mice; however assessment of left ventricular ejection fraction and fractional shortening revealed no evidence of cardiac dysfunction between groups. Furthermore, Cx40-/-, Panx1-/-, and Cx40-/-Panx1-/- mice demonstrated impaired endothelial-mediated vasodilation of aortic segments to increasing concentrations of methacholine (MCh) compared with WT, highlighting roles for both Cx40 and Panx1 in vascular endothelial cell (EC) function. Surprisingly, elevated kidney renin mRNA expression, plasma renin activity, and extraglomerular renin-producing cell populations found in Cx40-/- mice was further exaggerated in double knockout mice. Thus, while gestation and gross development were conserved in Cx40-/-Panx1-/- mice, they exhibit cardiac hypertrophy, hypertension, and impaired endothelial-mediated vasodilation that phenocopies Cx40-/- mice. Nevertheless, the augmented renin homeostasis observed in the double knockout mice suggests that both Cx40 and Panx1 may play an integrative role.
Collapse
Affiliation(s)
| | - Meghan Jelen
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Canada
| | - Kevin Barr
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Canada
| | - Leon J DeLalio
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, U.S.A
| | - Qingping Feng
- Department of Physiology and Pharmacology London, ON, Canada
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, U.S.A
| | - Robert Gros
- Department of Physiology and Pharmacology London, ON, Canada
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Dale W Laird
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Canada
- Department of Physiology and Pharmacology London, ON, Canada
| |
Collapse
|
15
|
Kristiansen SB, Skovsted GF, Berchtold LA, Radziwon-Balicka A, Dreisig K, Edvinsson L, Sheykhzade M, Haanes KA. Role of pannexin and adenosine triphosphate (ATP) following myocardial ischemia/reperfusion. SCAND CARDIOVASC J 2019; 52:340-343. [PMID: 30481075 DOI: 10.1080/14017431.2018.1552793] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVES The purinergic system has not been investigated in detail following ischemia/reperfusion (I/R) injury in the heart. In the present study, we focus on both release and response to extracellular adenosine triphosphate (ATP). Pannexin (Panx) channels have been shown to be involved in ATP release from myocytes and can activate P2X1 and P2Y2 receptors on the coronary artery. DESIGN We applied a well-characterized I/R model in rats, with 24 hours of reperfusion. Panx expression in the myocardial tissue was measured with quantitative polymerase chain reaction (qPCR) and flow cytometry. ATP release was detected in situ using luminescence and the vascular response to nucleotides determined in a wire myograph. RESULTS Here, we show that Panx expression is increased after experimental myocardial I/R, leading to an increase in extracellular ATP release, which could be inhibited by probenecid. Functional studies revealed that the P2Y2 receptor-dependent contraction is reduced in the coronary artery after I/R, which might be a response to the increased ATP levels. CONCLUSION We, therefore, conclude that the regulation of the arterial purinergic system minimizes coronary contractions following ischemia.
Collapse
Affiliation(s)
- Sarah Brøgger Kristiansen
- a Department of Clinical Experimental Research, Glostrup Research Institute , Copenhagen University Hospital, Rigshospitalet-Glostrup , Glostrup , Denmark.,b Department of Drug Design and Pharmacology, Faculty of Health Sciences , University of Copenhagen , Copenhagen , Denmark
| | - Gry Freja Skovsted
- a Department of Clinical Experimental Research, Glostrup Research Institute , Copenhagen University Hospital, Rigshospitalet-Glostrup , Glostrup , Denmark.,c Experimental Pharmacology and Toxicology, Section of Experimental Animal Models, Department of Veterinary and Animal Sciences , University of Copenhagen , Copenhagen , Denmark
| | - Lukas Adrian Berchtold
- d Department of Pediatrics, Glostrup Research Institute , Copenhagen University Hospital , Glostrup , Denmark.,e Center for Genomic Medicine , Copenhagen University Hospital , Glostrup , Denmark
| | - Aneta Radziwon-Balicka
- a Department of Clinical Experimental Research, Glostrup Research Institute , Copenhagen University Hospital, Rigshospitalet-Glostrup , Glostrup , Denmark
| | - Karin Dreisig
- a Department of Clinical Experimental Research, Glostrup Research Institute , Copenhagen University Hospital, Rigshospitalet-Glostrup , Glostrup , Denmark
| | - Lars Edvinsson
- a Department of Clinical Experimental Research, Glostrup Research Institute , Copenhagen University Hospital, Rigshospitalet-Glostrup , Glostrup , Denmark
| | - Majid Sheykhzade
- b Department of Drug Design and Pharmacology, Faculty of Health Sciences , University of Copenhagen , Copenhagen , Denmark
| | - Kristian Agmund Haanes
- a Department of Clinical Experimental Research, Glostrup Research Institute , Copenhagen University Hospital, Rigshospitalet-Glostrup , Glostrup , Denmark
| |
Collapse
|
16
|
Chen Z, He L, Li L, Chen L. The P2X7 purinergic receptor: An emerging therapeutic target in cardiovascular diseases. Clin Chim Acta 2018; 479:196-207. [PMID: 29366837 DOI: 10.1016/j.cca.2018.01.032] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/19/2018] [Accepted: 01/19/2018] [Indexed: 10/24/2022]
Abstract
The P2X7 purinergic receptor, a calcium permeable cationic channel, is activated by extracellular ATP. Most studies show that P2X7 receptor plays an important role in the nervous system diseases, immune response, osteoporosis and cancer. Mounting evidence indicates that P2X7 receptor is also associated with cardiovascular disease. For example, the P2X7 receptor activated by ATP can attenuate myocardial ischemia-reperfusion injury. By contrast, inhibition of P2X7 receptor decreases arrhythmia after myocardial infarction, prolongs cardiac survival after a long term heart transplant, alleviates the dilated cardiomyopathy and the autoimmune myocarditis process. The P2X7 receptor also mitigates vascular diseases including atherosclerosis, hypertension, thrombosis and diabetic retinopathy. This review focuses on the latest research on the role and therapeutic potential of P2X7 receptor in cardiovascular diseases.
Collapse
Affiliation(s)
- Zhe Chen
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, China
| | - Lu He
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, China
| | - Lanfang Li
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, China.
| | - Linxi Chen
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, China.
| |
Collapse
|
17
|
Xu J, Chen L, Li L. Pannexin hemichannels: A novel promising therapy target for oxidative stress related diseases. J Cell Physiol 2017; 233:2075-2090. [PMID: 28295275 DOI: 10.1002/jcp.25906] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 03/09/2017] [Indexed: 12/16/2022]
Abstract
Pannexins, which contain three subtypes: pannexin-1, -2, and -3, are vertebrate glycoproteins that form non-junctional plasma membrane intracellular hemichannels via oligomerization. Oxidative stress refers to an imbalance of the generation and elimination of reactive oxygen species (ROS). Studies have shown that elevated ROS levels are pivotal in the development of a variety of diseases. Recent studies indicate that the occurrence of these oxidative stress related diseases is associated with pannexin hemichannels. It is also reported that pannexins regulate the production of ROS which in turn may increase the opening of pannexin hemichannels. In this paper, we review recent researches about the important role of pannexin hemichannels in oxidative stress related diseases. Thus, pannexin hemichannels, novel therapeutic targets, hold promise in managing oxidative stress related diseases such as the tumor, inflammatory bowel diseases (IBD), pulmonary fibrosis, chronic obstructive pulmonary disease (COPD), cardiovascular disease, insulin resistance (IR), and neural degeneration diseases.
Collapse
Affiliation(s)
- Jin Xu
- Learning Key Laboratory for Pharmacoproteomics, Institute of Pharmacy and Pharmacology, University of South China, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, P. R. China
| | - Linxi Chen
- Learning Key Laboratory for Pharmacoproteomics, Institute of Pharmacy and Pharmacology, University of South China, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, P. R. China
| | - Lanfang Li
- Learning Key Laboratory for Pharmacoproteomics, Institute of Pharmacy and Pharmacology, University of South China, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, P. R. China
| |
Collapse
|
18
|
Orellana JA. Physiological Functions of Glial Cell Hemichannels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 949:93-108. [DOI: 10.1007/978-3-319-40764-7_5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
19
|
Ischemia triggered ATP release through Pannexin-1 channel by myocardial cells activates sympathetic fibers. Microvasc Res 2015; 104:32-7. [PMID: 26596404 DOI: 10.1016/j.mvr.2015.11.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 10/31/2015] [Accepted: 11/16/2015] [Indexed: 12/14/2022]
Abstract
The cardiovascular system is extensively innervated by the autonomic nervous system, and the autonomic modulation including sympathetic innervation is crucial to the function of heart during normal and ischemic conditions. Severe myocardial ischemia could cause acute myocardial infarction, which is one of the leading diseases in the world. Thus studying the sympathetic modulation during ischemia could reduce the probability of myocardial infarction and further heart failure. The neurotransmitter ATP is released by myocardial cells during ischemia; however, the effect of ATP release remains elusive. We examined whether ATP released during ischemia functions as a neurotransmitter that activates sympathetic nerve in the heart. A novel technique of recording the sympathetic fiber calcium imaging in mouse cardiac tissue slices was used. We have applied the Cre/loxP system to specifically express GCaMP3, a genetically encoded calcium indicator, in the sympathetic nerve. Using this technique, we found that ATP released by myocardial cells through Pannexin-1 channel during ischemia could evoke calcium responses in cardiac sympathetic nerve fibers. Our study provides a new approach to study the cell and nerve interaction in the cardiac system, as well as a new understanding of ATP function during ischemia.
Collapse
|
20
|
Advances in the pharmacology of lGICs auxiliary subunits. Pharmacol Res 2015; 101:65-73. [PMID: 26255765 DOI: 10.1016/j.phrs.2015.07.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 07/25/2015] [Accepted: 07/26/2015] [Indexed: 11/21/2022]
Abstract
Ligand-gated ion channels (LGICs) are cell surface integral proteins that mediate the fast neurotransmission in the nervous system. LGICs require auxiliary subunits for their trafficking, assembly and pharmacological modulation. Auxiliary subunits do not form functional homomeric receptors, but are reported to assemble with the principal subunits in order to modulate their pharmacological profiles. For example, nACh receptors are built at least by co-assemble of α and β subunits, and the neuronal auxiliary subunits β3 and α5 and muscle type β, δ, γ, and ϵ determine the agonist affinity of these receptors. Serotonergic 5-HT3B, 5-HT3C, 5-HT3D and 5-HT3E are reported to assemble with the 5-HT3A subunit to modulate its pharmacological profile. Functional studies evaluating the role of γ2 and δ auxiliary subunits of GABAA receptors have made important advances in the understanding of the action of benzodiazepines, ethanol and neurosteroids. Glycine receptors are composed principally by α1-3 subunits and the auxiliary subunit β determines their synaptic location and their pharmacological response to propofol and ethanol. NMDA receptors appear to be functional as heterotetrameric channels. So far, the existence of NMDA auxiliary subunits is controversial. On the other hand, Kainate receptors are modulated by NETO 1 and 2. AMPA receptors are modulated by TARPs, Shisa 9, CKAMP44, CNIH2-3 auxiliary proteins reported that controls their trafficking, conductance and gating of channels. P2X receptors are able to associate with auxiliary Pannexin-1 protein to modulate P2X7 receptors. Considering the pharmacological relevance of different LGICs auxiliary subunits in the present work we will highlight the therapeutic potential of these modulator proteins.
Collapse
|
21
|
Li L, He L, Wu D, Chen L, Jiang Z. Pannexin-1 channels and their emerging functions in cardiovascular diseases. Acta Biochim Biophys Sin (Shanghai) 2015; 47:391-6. [PMID: 25921414 DOI: 10.1093/abbs/gmv028] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 02/04/2015] [Indexed: 11/15/2022] Open
Abstract
Pannexin-1, Pannexin-2, and Pannexin-3 are three members of the Pannexin family of channel-forming glycoprotein. Their primary function is defined by their ability to form single-membrane channels. Pannexin-1 ubiquitously exists in many cells and organs throughout the body and is specially distributed in the circulatory system, while the expressions of Pannexin-2 and Pannexin-3 are mostly restricted to organs and tissues. Pannexin-1 oligomers have been shown to be functional single membrane channels that connect intracellular and extracellular compartments and are not intercellular channels in appositional membranes. The physiological functions of Pannexin-1 are to link to the adenosine triphosphate efflux that acts as a paracrine signal, and regulate cellular inflammasomes in a variety of cell types under physiological and pathophysiological conditions. However, there are still many functions to be explored. This review summarizes recent reports and discusses the role of Pannexin-1 in cardiovascular diseases, including ischemia, arrhythmia, cardiac fibrosis, and hypertension. Pannexin-1 has been suggested as an exciting, clinically relevant target in cardiovascular diseases.
Collapse
Affiliation(s)
- Lanfang Li
- Post-doctoral Mobile Stations for Basic Medicine, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang 421001, China Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, University of South China, Hengyang 421001, China
| | - Lu He
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, University of South China, Hengyang 421001, China
| | - Di Wu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, University of South China, Hengyang 421001, China
| | - Linxi Chen
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, University of South China, Hengyang 421001, China
| | - Zhisheng Jiang
- Post-doctoral Mobile Stations for Basic Medicine, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang 421001, China
| |
Collapse
|
22
|
Mahi N, Kumar A, Jaggi AS, Singh N, Dhawan R. Possible role of pannexin 1/P2x7 purinoceptor in neuroprotective mechanism of ischemic postconditioning in mice. J Surg Res 2015; 196:190-9. [DOI: 10.1016/j.jss.2015.02.050] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 02/18/2015] [Accepted: 02/19/2015] [Indexed: 01/01/2023]
|
23
|
Burnstock G, Pelleg A. Cardiac purinergic signalling in health and disease. Purinergic Signal 2015; 11:1-46. [PMID: 25527177 PMCID: PMC4336308 DOI: 10.1007/s11302-014-9436-1] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 11/25/2014] [Indexed: 01/09/2023] Open
Abstract
This review is a historical account about purinergic signalling in the heart, for readers to see how ideas and understanding have changed as new experimental results were published. Initially, the focus is on the nervous control of the heart by ATP as a cotransmitter in sympathetic, parasympathetic, and sensory nerves, as well as in intracardiac neurons. Control of the heart by centers in the brain and vagal cardiovascular reflexes involving purines are also discussed. The actions of adenine nucleotides and nucleosides on cardiomyocytes, atrioventricular and sinoatrial nodes, cardiac fibroblasts, and coronary blood vessels are described. Cardiac release and degradation of ATP are also described. Finally, the involvement of purinergic signalling and its therapeutic potential in cardiac pathophysiology is reviewed, including acute and chronic heart failure, ischemia, infarction, arrhythmias, cardiomyopathy, syncope, hypertrophy, coronary artery disease, angina, diabetic cardiomyopathy, as well as heart transplantation and coronary bypass grafts.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London, NW3 2PF, UK,
| | | |
Collapse
|
24
|
Kurtenbach S, Kurtenbach S, Zoidl G. Gap junction modulation and its implications for heart function. Front Physiol 2014; 5:82. [PMID: 24578694 PMCID: PMC3936571 DOI: 10.3389/fphys.2014.00082] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 02/10/2014] [Indexed: 01/04/2023] Open
Abstract
Gap junction communication (GJC) mediated by connexins is critical for heart function. To gain insight into the causal relationship of molecular mechanisms of disease pathology, it is important to understand which mechanisms contribute to impairment of gap junctional communication. Here, we present an update on the known modulators of connexins, including various interaction partners, kinases, and signaling cascades. This gap junction network (GJN) can serve as a blueprint for data mining approaches exploring the growing number of publicly available data sets from experimental and clinical studies.
Collapse
Affiliation(s)
- Stefan Kurtenbach
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| | - Sarah Kurtenbach
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada
| | - Georg Zoidl
- Department of Psychology, Faculty of Health, York University Toronto, ON, Canada ; Department of Biology, Faculty of Science, York University Toronto, ON, Canada ; Center for Vision Research, York University Toronto, ON, Canada
| |
Collapse
|
25
|
Isakson BE, Thompson RJ. Pannexin-1 as a potentiator of ligand-gated receptor signaling. Channels (Austin) 2014; 8:118-23. [PMID: 24576994 PMCID: PMC4048300 DOI: 10.4161/chan.27978] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Pannexins are a class of plasma membrane spanning proteins that presumably form a hexameric, non-selective ion channel. Although similar in secondary structure to the connexins, pannexins notably do not form endogenous gap junctions and act as bona fide ion channels. The pannexins have been primarily studied as ATP-release channels, but the overall diversity of their functions is still being elucidated. There is an intriguing theme with pannexins that has begun to develop. In this review we analyze several recent reports that converge on the idea that pannexin channels (namely Panx1) can potentiate ligand-gated receptor signaling. Although the literature remains sparse, this emerging concept appears consistent between both ionotropic and metabotropic receptors of several ligand families.
Collapse
Affiliation(s)
- Brant E Isakson
- Robert M. Berne Cardiovascular Research Center; University of Virginia School of Medicine; Charlottesville, VA USA; Department of Molecular Physiology and Biophysics; University of Virginia School of Medicine; Charlottesville, VA USA
| | - Roger J Thompson
- Hotchkiss Brain Institute; Department of Cell Biology and Anatomy; University of Calgary; Calgary, AB Canada
| |
Collapse
|
26
|
Abstract
The pannexins (Panxs) are a family of chordate proteins homologous to the invertebrate gap junction forming proteins named innexins. Three distinct Panx paralogs (Panx1, Panx2, and Panx3) are shared among the major vertebrate phyla, but they appear to have suppressed (or even lost) their ability to directly couple adjacent cells. Connecting the intracellular and extracellular compartments is now widely accepted as Panx's primary function, facilitating the passive movement of ions and small molecules along electrochemical gradients. The tissue distribution of the Panxs ranges from pervasive to very restricted, depending on the paralog, and are often cell type-specific and/or developmentally regulated within any given tissue. In recent years, Panxs have been implicated in an assortment of physiological and pathophysiological processes, particularly with respect to ATP signaling and inflammation, and they are now considered to be a major player in extracellular purinergic communication. The following is a comprehensive review of the Panx literature, exploring the historical events leading up to their discovery, outlining our current understanding of their biochemistry, and describing the importance of these proteins in health and disease.
Collapse
Affiliation(s)
- Stephen R Bond
- Genome Technology Branch, Division of Intramural Research, National Human Genome Research Institute, National Institutes of Health Bethesda, MD, USA ; Department of Cellular and Physiological Science, Life Sciences Institute, University of British Columbia Vancouver, BC, Canada
| | - Christian C Naus
- Department of Cellular and Physiological Science, Life Sciences Institute, University of British Columbia Vancouver, BC, Canada
| |
Collapse
|
27
|
Role of P2X7 purinoceptors in neuroprotective mechanism of ischemic postconditioning in mice. Mol Cell Biochem 2014; 390:161-73. [DOI: 10.1007/s11010-014-1967-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 01/21/2014] [Indexed: 01/23/2023]
|
28
|
Burnstock G, Ralevic V. Purinergic signaling and blood vessels in health and disease. Pharmacol Rev 2013; 66:102-92. [PMID: 24335194 DOI: 10.1124/pr.113.008029] [Citation(s) in RCA: 227] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Purinergic signaling plays important roles in control of vascular tone and remodeling. There is dual control of vascular tone by ATP released as a cotransmitter with noradrenaline from perivascular sympathetic nerves to cause vasoconstriction via P2X1 receptors, whereas ATP released from endothelial cells in response to changes in blood flow (producing shear stress) or hypoxia acts on P2X and P2Y receptors on endothelial cells to produce nitric oxide and endothelium-derived hyperpolarizing factor, which dilates vessels. ATP is also released from sensory-motor nerves during antidromic reflex activity to produce relaxation of some blood vessels. In this review, we stress the differences in neural and endothelial factors in purinergic control of different blood vessels. The long-term (trophic) actions of purine and pyrimidine nucleosides and nucleotides in promoting migration and proliferation of both vascular smooth muscle and endothelial cells via P1 and P2Y receptors during angiogenesis and vessel remodeling during restenosis after angioplasty are described. The pathophysiology of blood vessels and therapeutic potential of purinergic agents in diseases, including hypertension, atherosclerosis, ischemia, thrombosis and stroke, diabetes, and migraine, is discussed.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London NW3 2PF, UK; and Department of Pharmacology, The University of Melbourne, Australia.
| | | |
Collapse
|
29
|
Abstract
Endogenous nucleotides have widespread actions in the cardiovascular system, but it is only recently that the P2X and P2Y receptor subtypes, at which they act, have been identified and subtype-selective agonists and antagonists developed. These advances have greatly increased our understanding of the physiological and pathophysiological functions of P2X and P2Y receptors, but investigation of the clinical usefulness of selective ligands is at an early stage. Nonetheless, the evidence considered in this review demonstrates clearly that various cardiovascular disorders, including vasospasm, hypertension, congestive heart failure and cardiac damage during ischemic episodes, may be viable targets. With further development of novel, selective agonists and antagonists, our understanding will continue to improve and further therapeutic applications are likely to be discovered.
Collapse
|
30
|
Vessey DA, Li L, Imhof I, Honbo N, Karliner JS. FTY720 postconditions isolated perfused heart by a mechanism independent of sphingosine kinase 2 and different from S1P or ischemic postconditioning. Med Sci Monit Basic Res 2013; 19:126-32. [PMID: 23567658 PMCID: PMC3659128 DOI: 10.12659/msmbr.883877] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background We investigated the hypothesis that postconditioning by FTY720 (FTY) in isolated perfused mouse hearts is independent of the sphingosine 1-phosphate (S1P) pathway. Material/Methods Ex vivo hearts were exposed to postconditioning (POST) by either ischemia or FTY720. Protection against ischemia/reperfusion (IR) injury was measured by recovery of left ventricular developed pressure (LVDP) and infarct size. Results FTY effectively postconditioned (POST) ex vivo hearts against ischemia/reperfusion (IR) injury as measured by recovery of LVDP and a low infarct size. FTY protection, unlike S1P but like sphingosine (Sph), was insensitive to inhibition of S1P G-Protein Coupled Receptors (GPCRs) or inhibition of PI3 kinase. Protection by FTY and Sph was however blocked by inhibitors of PKA and PKG. Thus, FTY follows the same cardioprotective pathway as Sph. This was further supported by studies of FTY POST in knockout (KO) mice lacking the SphK2 form of Sph kinase that is needed for phosphorylation of FTY to an S1P analog. In the absence of SphK2, FTY (and Sph) POST was still cardioprotective. This differed from the effect of SphK2 KO on protection by ischemic POST (IPOST). IPOST was not effective in KO hearts. To see if the GPCR signaling pathway to protection is normal in KO hearts, we looked at POST by GPCR agonists S1P and adenosine. Both provided effective protection even in KO hearts suggesting that the problem with IPOST in KO hearts is a low level of S1P available for release during IPOST. Thus, pharmacologic POST with FTY or Sph, like adenosine and S1P, is unaffected in the KO. Conclusions FTY720 administered in vivo might behave in a dual manner showing both S1P-like effects and sphingosine-like effects. It appears that the latter may have been overlooked and may be the more important in aging hearts.
Collapse
Affiliation(s)
- Donald A Vessey
- Liver Study Unit, Veterans Affairs Medical Center, San Francisco, CA, USA.
| | | | | | | | | |
Collapse
|
31
|
Abstract
Purinergic receptors have attracted growing interest as therapeutic targets. This perspective focuses on P2X(4) receptors as a new cardioprotective target in heart failure.
Collapse
Affiliation(s)
- Ronghua Yang
- Calhoun Cardiovascular Center, University of Connecticut Health Center School of Medicine, Farmington, CT 06030, USA
| | | |
Collapse
|
32
|
Young CNJ, Brutkowski W, Lien CF, Arkle S, Lochmüller H, Zabłocki K, Górecki DC. P2X7 purinoceptor alterations in dystrophic mdx mouse muscles: relationship to pathology and potential target for treatment. J Cell Mol Med 2012; 16:1026-37. [PMID: 21794079 PMCID: PMC4365874 DOI: 10.1111/j.1582-4934.2011.01397.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal inherited muscle disorder. Pathological characteristics of DMD skeletal muscles include, among others, abnormal Ca(2+) homeostasis and cell signalling. Here, in the mdx mouse model of DMD, we demonstrate significant P2X7 receptor abnormalities in isolated primary muscle cells and cell lines and in dystrophic muscles in vivo. P2X7 mRNA expression in dystrophic muscles was significantly up-regulated but without alterations of specific splice variant patterns. P2X7 protein was also up-regulated and this was associated with altered function of P2X7 receptors producing increased responsiveness of cytoplasmic Ca(2+) and extracellular signal-regulated kinase (ERK) phosphorylation to purinergic stimulation and altered sensitivity to NAD. Ca(2+) influx and ERK signalling were stimulated by ATP and BzATP, inhibited by specific P2X7 antagonists and insensitive to ivermectin, confirming P2X7 receptor involvement. Despite the presence of pannexin-1, prolonged P2X7 activation did not trigger cell permeabilization to propidium iodide or Lucifer yellow. In dystrophic mice, in vivo treatment with the P2X7 antagonist Coomassie Brilliant Blue reduced the number of degeneration-regeneration cycles in mdx skeletal muscles. Altered P2X7 expression and function is thus an important feature in dystrophic mdx muscle and treatments aiming to inhibit P2X7 receptor might slow the progression of this disease.
Collapse
Affiliation(s)
- Christopher N J Young
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | | | | | | | | | | | | |
Collapse
|
33
|
Cosentino S, Banfi C, Burbiel JC, Luo H, Tremoli E, Abbracchio MP. Cardiomyocyte death induced by ischaemic/hypoxic stress is differentially affected by distinct purinergic P2 receptors. J Cell Mol Med 2012; 16:1074-84. [PMID: 21762374 PMCID: PMC4365886 DOI: 10.1111/j.1582-4934.2011.01382.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Blood levels of extracellular nucleotides (e.g. ATP) are greatly increased during heart ischaemia, but, despite the presence of their specific receptors on cardiomyocytes (both P2X and P2Y subtypes), their effects on the subsequent myocardial damage are still unknown. In this study, we aimed at investigating the role of ATP and specific P2 receptors in the appearance of cell injury in a cardiac model of ischaemic/hypoxic stress. Cells were maintained in a modular incubator chamber in a controlled humidified atmosphere of 95% N2 for 16 hrs in a glucose-free medium. In this condition, we detected an early increase in the release of ATP in the culture medium, which was followed by a massive increase in the release of cytoplasmic histone-associated-DNA-fragments, a marker of apoptosis. Addition of either apyrase, which degrades extracellular ATP, or various inhibitors of ATP release via connexin hemichannels fully abolished ischaemic/hypoxic stress-associated apoptosis. To dissect the role of specific P2 receptor subtypes, we used a combined approach: (i) non-selective and, when available, subtype-selective P2 antagonists, were added to cardiomyocytes before ischaemic/hypoxic stress; (ii) selected P2 receptors genes were silenced via specific small interfering RNAs. Both approaches indicated that the P2Y2 and P2χ7 receptor subtypes are directly involved in the induction of cell death during ischaemic/hypoxic stress, whereas the P2Y4 receptor has a protective effect. Overall, these findings indicate a role for ATP and its receptors in modulating cardiomyocyte damage during ischaemic/hypoxic stress.
Collapse
|
34
|
Baroja-Mazo A, Barberà-Cremades M, Pelegrín P. The participation of plasma membrane hemichannels to purinergic signaling. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1828:79-93. [PMID: 22266266 DOI: 10.1016/j.bbamem.2012.01.002] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 12/30/2011] [Accepted: 01/04/2012] [Indexed: 12/17/2022]
Abstract
The field of hemichannels is closely related to the purinergic signaling and both areas have been growing in parallel. Hemichannels open in response to a wide range of stressful conditions, such as ischemia, pressure or swelling. Hemichannels represent an important mechanism for the cellular release of adenosine 5'-triphosphate (ATP), which is an agonist of the P2Y and P2X family of purinergic receptors. Therefore, hemichannels are key molecules in the regulation of purinergic receptor activation, during physiological and pathophysiological conditions. Furthermore, purinergic receptor activation can also lead to the opening of hemichannels and the subsequent amplification of purinergic signaling via a positive signaling feedback loop, giving rise to the concept of ATP-induced ATP release. Purinergic receptor signaling is involved in regulating many physiological and pathophysiological processes. P2Y receptors activate inositol trisphosphate and transiently increase intracellular calcium. This signaling opens both connexin and pannexin channels, therefore contributing to the expansion of calcium waves across astrocytes and epithelial cells. In addition, several of the P2X receptor subtypes, including the P2X2, P2X4 and P2X7 receptors, activate select cellular permeation pathways to large molecules, including the pannexin-1 channels, which are involved in the initiation of inflammatory responses and cell death. Consequently, the interplay between purinergic receptors and hemichannels could represent a novel target with substantial therapeutic implications in areas such as chronic pain, inflammation or atherosclerosis. This article is part of a Special Issue entitled: The communicating junctions, roles and dysfunctions.
Collapse
Affiliation(s)
- Alberto Baroja-Mazo
- University Hospital Virgen de la Arrixaca, Fundación Formación Investigación Sanitaria Región Murcia, Murcia, Spain
| | | | | |
Collapse
|
35
|
Hervé JC, Derangeon M, Sarrouilhe D, Giepmans BNG, Bourmeyster N. Gap junctional channels are parts of multiprotein complexes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2011; 1818:1844-65. [PMID: 22197781 DOI: 10.1016/j.bbamem.2011.12.009] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 11/28/2011] [Accepted: 12/06/2011] [Indexed: 12/16/2022]
Abstract
Gap junctional channels are a class of membrane channels composed of transmembrane channel-forming integral membrane proteins termed connexins, innexins or pannexins that mediate direct cell-to-cell or cell-to extracellular medium communication in almost all animal tissues. The activity of these channels is tightly regulated, particularly by intramolecular modifications as phosphorylations of proteins and via the formation of multiprotein complexes where pore-forming subunits bind to auxiliary channel subunits and associate with scaffolding proteins that play essential roles in channel localization and activity. Scaffolding proteins link signaling enzymes, substrates, and potential effectors (such as channels) into multiprotein signaling complexes that may be anchored to the cytoskeleton. Protein-protein interactions play essential roles in channel localization and activity and, besides their cell-to-cell channel-forming functions, gap junctional proteins now appear involved in different cellular functions (e.g. transcriptional and cytoskeletal regulations). The present review summarizes the recent progress regarding the proteins capable of interacting with junctional proteins and highlights the function of these protein-protein interactions in cell physiology and aberrant function in diseases. This article is part of a Special Issue entitled: The Communicating junctions, composition, structure and functions.
Collapse
Affiliation(s)
- Jean-Claude Hervé
- Institut de Physiologie et Biologie Cellulaires, Université de Poitiers, CNRS, Poitiers, France.
| | | | | | | | | |
Collapse
|
36
|
Poornima V, Madhupriya M, Kootar S, Sujatha G, Kumar A, Bera AK. P2X7 receptor-pannexin 1 hemichannel association: effect of extracellular calcium on membrane permeabilization. J Mol Neurosci 2011; 46:585-94. [PMID: 21932038 DOI: 10.1007/s12031-011-9646-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Accepted: 08/30/2011] [Indexed: 10/17/2022]
Abstract
Activation of P2X(7) receptor (P2X(7)R) and pannexin have been implicated in membrane permeabilization associated with ischemic cell death and many other inflammatory processes. P2X(7)R has a unique property of forming large pore upon repeated or prolonged application of agonist like ATP or 2', 3'-(4-benzoyl) benzoyl ATP. It has been proposed that pannexin 1 (panx1) hemichannel associates with P2X(7)R to form large pore, though the actual mechanism is not yet understood. Calcium concentration in extracellular milieu drops in many patho-physiological conditions, e.g. ischemia, when P2X(7)R/pannexin is also known to be activated. Therefore, we hypothesize that extracellular calcium ([Ca(2+)](o)) plays an important role in the coupling of P2X(7)R-panx1 and subsequent membrane permeabilization. In this study we show that membrane permeability of the P2X(7)R and panx1 expressing N2A cell increases in ([Ca(2+)](o))-free solution. In [Ca(2+)](o)-free solution, fluorescent dye calcein trapped cells exhibited time-dependent dye leakage resulting in about 50% decrease of fluorescence intensity in 30 min. Control cells in 2 mM [Ca(2+)](o) did not show such leakage. Like N2A cells, mixed culture of neuron and glia, derived from hippocampal progenitor cells showed similar dye leakage. Dye leakage was blocked either by pannexin-specific blocker, carbenoxolone or P2X(7)R antagonists, Brilliant Blue G, and oxidized ATP. Furthermore P2X(7)R and panx1 were co-immunoprecipitated. The amount of P2X(7)R protein pulled-down with panx1, increased by twofold when cells were incubated 30 min in [Ca(2+)](o)-free buffer. Taken together, the results of this study demonstrate the activation and association of P2X(7)R-panx1, triggered by the removal of [Ca(2+)](o).
Collapse
Affiliation(s)
- V Poornima
- Department of Biotechnology, IIT Madras, Chennai, 600036, India
| | | | | | | | | | | |
Collapse
|
37
|
Rodríguez-Sinovas A, Sánchez JA, Fernandez-Sanz C, Ruiz-Meana M, Garcia-Dorado D. Connexin and pannexin as modulators of myocardial injury. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2011; 1818:1962-70. [PMID: 21839721 DOI: 10.1016/j.bbamem.2011.07.041] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 07/20/2011] [Accepted: 07/28/2011] [Indexed: 01/02/2023]
Abstract
Multicellular organisms have developed a variety of mechanisms that allow communication between their cells. Whereas some of these systems, as neurotransmission or hormones, make possible communication between remote areas, direct cell-to-cell communication through specific membrane channels keep in contact neighboring cells. Direct communication between the cytoplasm of adjacent cells is achieved in vertebrates by membrane channels formed by connexins. However, in addition to allowing exchange of ions and small metabolites between the cytoplasms of adjacent cells, connexin channels also communicate the cytosol with the extracellular space, thus enabling a completely different communication system, involving activation of extracellular receptors. Recently, the demonstration of connexin at the inner mitochondrial membrane of cardiomyocytes, probably forming hemichannels, has enlarged the list of actions of connexins. Some of these mechanisms are also shared by a different family of proteins, termed pannexins. Importantly, these systems allow not only communication between healthy cells, but also play an important role during different types of injury. The aim of this review is to discuss the role played by both connexin hemichannels and pannexin channels in cell communication and injury. This article is part of a Special Issue entitled: The Communicating junctions, composition, structure and characteristics.
Collapse
|
38
|
Vessey DA, Li L, Kelley M. P2X7 receptor agonists pre- and postcondition the heart against ischemia-reperfusion injury by opening pannexin-1/P2X₇ channels. Am J Physiol Heart Circ Physiol 2011; 301:H881-7. [PMID: 21685263 DOI: 10.1152/ajpheart.00305.2011] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Protection of the heart from ischemia-reperfusion injury can be achieved by ischemic preconditioning and ischemic postconditioning. Previous studies revealed that a complex of pannexin-1 with the P2X(7) receptor forms a channel during ischemic preconditioning and ischemic postconditioning that results in the release of endogenous cardioprotectants. ATP binds to P2X(7) receptors, inducing the formation of a channel in association with pannexin-1. We hypothesized that this channel would provide a pathway for the release of these same cardioprotectants. Preconditioning-isolated perfused rat hearts with 0.4 μM ATP preceding 40 min of ischemia minimized infarct size upon subsequent reperfusion (5% of risk area) and resulted in >80% recovery of left ventricular developed pressure. Postconditioning with ATP after ischemia during reperfusion was also protective (6% infarct and 72% recovery of left ventricular developed pressure). Antagonists of both pannexin-1 (carbenoxolone and mefloquine) and P2X(7) receptors (brilliant blue G and A438079) blocked ATP pre- and postconditioning, indicating that ATP protection was elicited via the opening of a pannexin-1/P2X(7) channel. An antagonist of binding of the endogenous cardioprotectant sphingosine 1-phosphate to its G protein-coupled receptor diminished protection by ATP, which is also consistent with an ATP-dependent release of cardioprotectants. Suramin, an antagonist of binding of ATP (and ADP) to P2Y receptors, was without effect on ATP protection. Benzoyl benzoyl-ATP, a more specific P2X(7) agonist, was also a potent pre- and postconditioning agent and sensitive to blockade by pannexin-1/P2X(7) channel antagonists. The data point out for the first time the potential of P2X(7) agonists as cardioprotectants.
Collapse
Affiliation(s)
- Donald A Vessey
- Liver Study Unit, Department of Veterans Affairs Medical Center, San Francisco, California 94121, USA.
| | | | | |
Collapse
|
39
|
A sphingosine kinase form 2 knockout sensitizes mouse myocardium to ischemia/reoxygenation injury and diminishes responsiveness to ischemic preconditioning. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2011; 2011:961059. [PMID: 21904650 PMCID: PMC3166792 DOI: 10.1155/2011/961059] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Accepted: 01/10/2011] [Indexed: 02/07/2023]
Abstract
Sphingosine kinase (SphK) exhibits two isoforms, SphK1 and SphK2. Both forms catalyze the synthesis of sphingosine 1-phosphate (S1P), a sphingolipid involved in ischemic preconditioning (IPC). Since the ratio of SphK1:SphK2 changes dramatically with aging, it is important to assess the role of SphK2 in IR injury and IPC. Langendorff mouse hearts were subjected to IR (30 min equilibration, 50 min global ischemia, and 40 min reperfusion). IPC consisted of 2 min of ischemia and 2 min of reperfusion for two cycles. At baseline, there were no differences in left ventricular developed pressure (LVDP), ± dP/dtmax, and heart rate between SphK2 null (KO) and wild-type (WT) hearts. In KO hearts, SphK2 activity was undetectable, and SphK1 activity was unchanged compared to WT. Total SphK activity was reduced by 53%. SphK2 KO hearts subjected to IR exhibited significantly more cardiac damage (37 ± 1% infarct size) compared with WT (28 ± 1% infarct size); postischemic recovery of LVDP was lower in KO hearts. IPC exerted cardioprotection in WT hearts. The protective effect of IPC against IR was diminished in KO hearts which had much higher infarction sizes (35 ± 2%) compared to the IPC/IR group in control hearts (12 ± 1%). Western analysis revealed that KO hearts had substantial levels of phosphorylated p38 which could predispose the heart to IR injury. Thus, deletion of the SphK2 gene sensitizes the myocardium to IR injury and diminishes the protective effect of IPC.
Collapse
|
40
|
Vessey DA, Li L, Kelley M. Ischemic preconditioning requires opening of pannexin-1/P2X7 channels not only during preconditioning but again after index ischemia at full reperfusion. Mol Cell Biochem 2011; 351:77-84. [DOI: 10.1007/s11010-011-0713-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Accepted: 01/04/2011] [Indexed: 10/18/2022]
|
41
|
Kienitz MC, Bender K, Dermietzel R, Pott L, Zoidl G. Pannexin 1 constitutes the large conductance cation channel of cardiac myocytes. J Biol Chem 2010; 286:290-8. [PMID: 21041301 DOI: 10.1074/jbc.m110.163477] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
A large conductance (∼300 picosiemens) channel (LCC) of unknown molecular identity, activated by Ca(2+) release from the sarcoplasmic reticulum, particularly when augmented by caffeine, has been described previously in isolated cardiac myocytes. A potential candidate for this channel is pannexin 1 (Panx1), which has been shown to form large ion channels when expressed in Xenopus oocytes and mammalian cells. Panx1 function is implicated in ATP-mediated auto-/paracrine signaling, and a crucial role in several cell death pathways has been suggested. Here, we demonstrate that after culturing for 4 days LCC activity is no longer detected in myocytes but can be rescued by adenoviral gene transfer of Panx1. Endogenous LCCs and those related to expression of Panx1 share key pharmacological properties previously used for identifying and characterizing Panx1 channels. These data demonstrate that Panx1 constitutes the LCC of cardiac myocytes. Sporadic openings of single Panx1 channels in the absence of Ca(2+) release can trigger action potentials, suggesting that Panx1 channels potentially promote arrhythmogenic activities.
Collapse
Affiliation(s)
- Marie-Cecile Kienitz
- Department of Cellular Physiology, Ruhr-University Bochum, 44780 Bochum, Germany.
| | | | | | | | | |
Collapse
|
42
|
Barth K, Pfleger C, Linge A, Sim JA, Surprenant A, Steinbronn N, Strasser RH, Kasper M. Increased P2X7R expression in atrial cardiomyocytes of caveolin-1 deficient mice. Histochem Cell Biol 2010; 134:31-8. [DOI: 10.1007/s00418-010-0716-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2010] [Indexed: 02/06/2023]
|